Compositions and methods for treating Parkinson's Disease (PD) and related disorders

文档序号:1471274 发布日期:2020-02-21 浏览:9次 中文

阅读说明:本技术 治疗帕金森氏病(pd)和相关疾病的组合物和方法 (Compositions and methods for treating Parkinson's Disease (PD) and related disorders ) 是由 T.J.博罗迪 于 2018-04-04 设计创作,主要内容包括:本公开属于适于治疗哺乳动物疾病的药物组合物领域。本公开提供了用于治疗PD和相关疾病的包含非致病粪便微生物或无菌粪便滤液的新型组合物。本公开还提供了用本文公开的组合物治疗受试者的方法。(The present disclosure is in the field of pharmaceutical compositions suitable for treating diseases in mammals. The present disclosure provides novel compositions comprising non-pathogenic fecal microorganisms or sterile fecal filtrate for the treatment of PD and related diseases. The present disclosure also provides methods of treating a subject with the compositions disclosed herein.)

1. A method of treating Parkinson's Disease (PD) in a human subject in need thereof, said method comprising orally administering to said human subject a pharmaceutically active dose of a therapeutic composition comprising a live non-pathogenic fecal bacterium or a non-cellular fecal filtrate.

2. The method of claim 1, wherein the subject has a clinical stage of PD selected from stage I to stage V.

3. The method of claim 1, wherein the therapeutic composition comprises an unselected fecal microbiota.

4. The method of claim 2, wherein the fecal microbiota comprises a complete fecal microbiota of a donor.

5. The method of claim 2, wherein the fecal microbiota is from synthetic fecal material.

6. The method of claim 1, wherein the method reduces the rate of progression of PD in the subject.

7. The method of claim 1, wherein the dose is administered at least once daily for at least 2 weeks.

8. The method of claim 1, wherein the dose is administered at least twice weekly for at least two weeks.

9. The method of claim 7, wherein the dose is administered at least twice weekly for at least 4 weeks.

10. The method of claim 7, wherein the dose is administered at least twice weekly for at least 8 weeks.

11. The method of claim 1, wherein the dose is administered at least three times per week for at least 4 weeks.

12. The method of claim 1, wherein the therapeutic composition comprises both live non-pathogenic fecal bacteria and a non-cellular fecal filtrate.

13. The method of claim 1, wherein the method comprises a first dosing regimen followed by a second dosing regimen, wherein the second dosing regimen comprises a maintenance dose that is less than or equal to the dose of the first dosing regimen.

14. The method of claim 1, wherein the therapeutic composition is formulated for delayed or gradual enteric release.

15. The method of claim 1, wherein the therapeutic composition is formulated as an enteric capsule or an acid resistant capsule.

16. The method of claim 1, wherein the pharmaceutically active dose is 108-1014cfu or total number of cells.

17. The method of claim 1, wherein the therapeutic composition is in liquid, frozen, freeze-dried, spray-dried, foam-dried, or powder form.

18. The method of claim 1, wherein the therapeutic composition comprises a cryoprotectant selected from the group consisting of polyethylene glycol, skim milk, erythritol, arabitol, sorbitol, glucose, fructose, alanine, glycine, proline, sucrose, lactose, ribose, trehalose, dimethyl sulfoxide (DMSO), glycerol, or a combination thereof.

19. The method of claim 1, wherein the subject is pre-treated with an antibiotic prior to administration of the composition.

20. The method of claim 1, wherein the method eliminates or reduces one or more symptoms of PD selected from tremor, slowed movement (bradykinesia), muscle stiffness, impaired posture and balance, spontaneous loss of movement, changes in speech, and changes in writing.

Technical Field

The present disclosure relates to pharmaceutical compositions and methods suitable for treating Parkinson's Disease (PD) and related diseases.

Background

The Gastrointestinal (GI) tract of mammals contains a variety of microorganisms. The interactions between these microorganisms and between the microorganisms and the host (e.g., the host immune system) form a microbial population. The healthy microflora provides a host with a number of benefits, including resistance to colonization by a broad spectrum of pathogens, biosynthesis and absorption of essential nutrients, and immune stimulation to maintain healthy intestinal epithelium and properly controlled systemic immunity. Unbalanced microflora (also referred to as "dysbiosis" or disrupted symbiosis) may lose their function and be more susceptible to infection by pathogens, altered metabolic characteristics, or to elicit pro-inflammatory signals that can lead to local or systemic inflammation or autoimmunity. In addition, this disrupted microbial population may be infected by one or more foreign pathogens, which may result in symptoms of pain, diarrhea, flatulence, constipation, and the like. Thus, the gut microbiota plays an important role in the pathogenesis of many diseases such as pathogenic infections of the gut.

The implantation or administration of human colonic microbiota into the patient's intestinal tract is known as Fecal Microbiota Transplantation (FMT), also commonly referred to as fecal bacterial therapy. It is believed that FMT can repopulate the gut with a variety of microorganisms that control the primary pathogen by creating an ecological environment that is not conducive to proliferation and survival of the primary pathogen. It represents a therapeutic regimen that enables rapid reconstitution of normal constitutive and functional intestinal microflora.

FMT has been used to treat Clostridium Difficile Infection (CDI). FMT has also been proposed for the treatment of other sources of intestinal infection, such as E.coli and vancomycin-resistant enterococci (VRE). It requires injection through a colonoscope, enema or nasojejunal tube of human microbiota in the form of homogenized stool or cultured stool components such as clostridia to be implanted into the colon to replace or eradicate pathogenic bacteria such as clostridium difficile.

Parkinson's Disease (PD) is a chronic progressive neurological disease that affects movement. PD develops with the loss of neurons in the substantia nigra region of the brain, resulting in a decrease in the availability of the neurotransmitter dopamine. Dopamine levels in the intestine may also decrease. Nearly one million people in the united states suffer from PD. PD usually occurs around 60 years of age and the risk of disease increases with age. Men are more susceptible to PD than women. Family history of PD and continued exposure to herbicides and pesticides may increase the risk of disease.

PD causes stiffness or slowness of movement of the limb. The progression of PD can be very slow, usually beginning with mild hand tremor or gentle speech or vague. Patients may also experience postural instability. Gastrointestinal symptoms include nausea or bloating, difficulty defecation, and constipation.

The severity of PD can be classified as mild, moderate or severe. Patients with mild PD develop tremors, changes in facial expression and possibly mild motor symptoms. During the moderate phase, the patient may exhibit bilateral motor symptoms, including slowed motion, difficulty balancing, and a foot feeling "stuck" on the ground. In the late stage of PD, walking becomes difficult and hallucinations and delusions become severe.

There is currently no cure for PD, but there are treatment regimens that can control the symptoms. Some drugs used to treat PD increase or replace dopamine in the human brain. The brain area can be regulated and symptoms improved by surgery. Although current treatments may alleviate some of the symptoms of PD, they are not as effective and are associated with side effects. Furthermore, over time, patients often experience a diminished response to these therapies. Thus, there is an unmet medical need for drugs or therapies that are more effective in treating PD and related diseases, and that are easier to administer.

Disclosure of Invention

The present disclosure provides compositions, methods and dosing regimens for treating or preventing PD.

In one aspect, the present disclosure provides a method of treating PD in a subject in need of treatment, wherein the method comprises administering to the subject a pharmaceutically active dose of a therapeutic composition comprising or derived from live non-pathogenic fecal bacteria or sterile fecal filtrate. In one aspect, the sterile fecal filtrate is derived from donor feces. In another aspect, the sterile fecal filtrate is derived from a cultured microorganism.

In another aspect, the present disclosure provides the use of a composition comprising live, non-pathogenic fecal bacteria in the manufacture of a medicament for the treatment of PD.

In one aspect, the present disclosure provides a method of treating PD in a subject in need of treatment, wherein the method comprises orally administering to the subject a pharmaceutically active dose of a therapeutic composition comprising a mixture of live non-pathogenic synthetic bacteria or a live non-pathogenic purified or extracted fecal microbiota, wherein the dose is administered on a dosing schedule of at least once or twice daily or at least once or twice weekly for at least three, eight, ten, or twenty consecutive weeks.

In one aspect, the present disclosure provides a method of treating PD in a subject in need of treatment, wherein the method comprises orally administering to the subject a pharmaceutically active dose of a therapeutic composition comprising a liquid, frozen, lyophilized, or encapsulated sterile fecal filtrate, wherein the dose is administered on a dosing schedule of at least once or twice daily or at least once or twice weekly for at least three, eight, ten, or twenty consecutive weeks.

In one aspect, a method achieves a PD remission rate, cure rate, response rate, or regression rate of at least about 80%. In one aspect, the present disclosure provides a method of eliminating or reducing one or more symptoms of PD selected from tremor, slowed movement (bradykinesia), muscle stiffness, impaired posture and balance, loss of spontaneous movement, changes in speech, and changes in writing.

In one aspect, the fecal microbiota in the therapeutic composition includes substantially intact and unselected fecal microbiota, recombinant fecal material, synthetic fecal material of the donor.

Detailed Description

Unless otherwise defined herein, terms will be understood by those of ordinary skill in the relevant art in light of conventional usage.

The term "treating" as used herein refers to (i) completely or partially inhibiting a disease, disorder or condition, e.g., arresting its development; (ii) a complete or partial remission of the disease, disorder or condition, e.g., regression of the disease, disorder and/or condition; or (iii) completely or partially preventing the disease, disorder or condition from occurring in a patient who may be predisposed to the disease, disorder and/or condition but has not yet been diagnosed as having the disease, disorder and/or condition. Likewise, "treatment" includes both actual therapeutic and prophylactic measures.

As used herein, a "therapeutically effective amount" or "pharmaceutically active dose" refers to an amount of a composition effective to treat a given disease, disorder, or condition.

As used herein, "microbiota" and "flora" refer to a microflora that persists and transiently lives in or on a subject, including eukaryotes, archaea, bacteria, and viruses (including bacterial viruses, i.e., bacteriophages). By unselected fecal microbiota is meant an unselected fecal microbiota or mixture derived from a donor fecal sample that is substantially similar to the microbial composition and population structure found in such fecal sample.

As used herein, "sterile fecal filtrate" or "non-cellular fecal filtrate" refers to the liquid component of fecal matter, wherein the liquid component is free or substantially free of cell-based living organisms (e.g., bacteria, fungi, or spores thereof), but retains bacteriophage and non-cellular biological matter. Preferably, the acellular or sterile fecal filtrate is also free of viruses of eukaryotic host cells.

As used herein, "remission rate, cure rate, or regression rate" refers to the percentage of patients who cured or had a remission or complete regression of the condition in response to a given treatment. Remission, cure, or regression of PD refers to complete cessation of one or more symptoms exhibited by the patient prior to receiving treatment as described herein. These symptoms may include, but are not limited to, tremors, slowed motion (bradykinesia), muscle stiffness, impaired posture and balance, loss of spontaneous movement, changes in speech, and changes in writing. Each of the above symptoms will be further explained below.

Tremor: tremor, or trembling, usually begins in a limb, usually the patient's hand or finger. The patient may rub his thumb and forefinger back and forth, known as "Marble tremor". One characteristic of parkinson's disease is that the patient's hands are tremor when relaxed (at rest).

Slowing of locomotion (bradykinesia): over time, parkinson's disease may reduce the mobility of the patient, slowing the patient's rate of activity, making simple things difficult and time consuming. The patient may have a shortened step while walking or may have difficulty getting off the chair. In addition, when the patient tries to walk, the patient may have a foot to wade, which is difficult to move.

Muscle stiffness: muscle stiffness may occur at any part of the patient's body. Stiff muscles can limit the range of motion of the patient and cause pain.

Impaired posture and balance: the patient may become hunched or the patient may experience balance problems due to parkinson's disease.

Loss of spontaneous motility: parkinson's disease patients may have a reduced ability to perform involuntary movements, including blinking, smiling or swinging the arms while walking.

Speech variation: parkinson's disease may cause speech impairment in patients. The patient may hesitate to speak softly, quickly, vaguely, or before opening. The speech intonation may tend to be single rather than normal.

Writing changes: writing becomes difficult and the patient's writing style may become very small.

As used herein, "response rate" refers to the percentage of patients who respond positively (e.g., a decrease in the severity or frequency of one or more symptoms) to a given treatment.

As used herein, the staging criteria for PD patients are the Hoehn and Yahr stages summarized in Table 1 (see also Hoehn Master and Yahr Ph, onset, progression and mortality of Parkinson's syndrome, Neurology1967,17: 427-42).

Table 1: hoehn and Yahr staging of Parkinson's disease.

Figure BDA0002304268610000041

As used herein, the "parkinson's disease composite score scale" or "UPDRS" refers to a scoring tool that tracks the longitudinal progression of PD, representing the level or severity of PD symptoms (table 2). UPDRS consists of a three-part test, including (1) mental, behavioral, and emotional, (2) Activities of Daily Living (ADL), and (3) motor parts. Part I, part II and part III tests included a total of 31 items. The score for each item ranged from 0 to 4, where 0 represents no damage and 4 represents the highest degree of damage. The sum of part I, part II and part III of each study visit was the UPDRS total score. The total score may reach 199. 199 represents the most severe (total) disability, 0-no disability. UPDRS is used to measure changes in efficacy variables from baseline during treatment as described herein.

Table 2: the parkinsonian composite score scale (UPDRS) which describes the severity of parkinson's disease.

Figure BDA0002304268610000052

Figure BDA0002304268610000061

Figure BDA0002304268610000071

Figure BDA0002304268610000101

Figure BDA0002304268610000111

Figure BDA0002304268610000121

Figure BDA0002304268610000131

Figure BDA0002304268610000141

Figure BDA0002304268610000151

As used herein, "symptomatic anti-parkinson's disease therapy" refers to a therapy based on the non-fecal microbiome. Examples of such therapy include any of bromocriptine, phenytoin, levodopa, ropinirole, pramipexole, rotigotine, cabergoline, entacapone, tolcapone, amantadine, and selegiline.

As used herein, "reducing the rate of progression of PD" refers to reducing the exacerbation experienced by a PD patient as compared to the exacerbation experienced by a PD patient who has not received the treatment provided herein for a period of time, e.g., as quantified by the UPDRS score.

As used herein, "delaying the need for symptomatic anti-parkinson's disease therapy" refers to delaying the need for symptomatic anti-parkinson's disease therapy in a PD patient receiving fecal microbiome based therapy as compared to a patient not receiving fecal microbiome based therapy.

As used herein, "early symptoms of PD" refers to one or more of:

(a) 4-8Hz Marble-like tremor occurs when one hand is at rest;

(b) the tremor is maximal when the patient is static, weakened when the patient moves, and disappeared when the patient sleeps;

(c) stiffness and slowed movement (bradycardia), reduced movement (hypokinesia) and difficulty in initiating movement (akinesia);

(d) facemasks, open mouth, and reduced blinking, which may be confused with depression;

(e) the posture becomes hunched back;

(f) starting walking is difficult; the foot is dragged to walk, the step pitch is small, and the arm is bent and kept at the waist without swinging along with the step;

(g) occasionally, footsteps are inadvertently accelerated, and patients occasionally run up to avoid falls (panicked gait);

(h) the center of gravity tends to fall forward (forward stroke) or backward (backward forward stroke) when moving, due to loss of postural reflexes;

(i) the voice becomes deep and has the characteristics of monotonous dysarthria and stuttering;

(j) hypokinesia and impaired control of distal musculature, resulting in too small fonts and increased difficulty in activities of daily life;

(k) infrequent blinking and lack of facial expressions;

(l) A reduction in motion;

(m) impaired postural reflexes; and/or

(n) gait feature abnormalities.

As used herein, an "early stage PD patient" is a PD patient in stage I or II Parkinson's disease as defined by Hoehn and Yahr, who does not require symptomatic anti-Parkinson's disease therapy. Preferably, such PD patients do not require symptomatic therapy for at least the next 9 months. Early stage PD patients can be identified by performing relevant tests.

As used herein, "eukaryotic" refers to cells that belong to the class consisting of nuclei and membrane-bound organelles.

As used herein, "bacterium" and "archaebacteria" refer to unicellular prokaryotes that lack membrane-bound nuclei and organelles.

As used herein, "colony forming units" (cfu) refers to an estimate of the number of viable microbial cells in a given sample.

As used herein, "live" means possessing proliferative capacity.

As used herein, "fecal bacteria" refers to bacteria that may be found in fecal material.

As used herein, "isolated" or "purified" means that a bacterium or other entity or substance has been (1) isolated from at least some of the components with which it was originally produced (whether in nature or in an experimental setting), and/or (2) produced, prepared, purified, and/or manufactured by the human hand. The isolated or purified bacteria can be separated from at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or more of the other components with which they are originally associated.

"cytotoxic" activity or bacteria, as used herein, includes the ability to kill bacterial cells, such as pathogenic bacterial cells. "cytostatic" activity or bacteria includes the ability to partially or fully inhibit the growth, metabolism and/or proliferation of bacterial cells, such as pathogenic bacterial cells.

The terms "pathogen" and "pathogenic" bacteria or any other organism or entity as used herein include any such organism or entity that is capable of causing or affecting a disease, disorder or condition of a host organism containing the organism or entity.

As used herein, a "spore" or "spore" population includes bacteria (or other unicellular organisms) that are generally viable, more resistant to environmental influences, such as heat and bactericides, than propagules of the same bacteria, and are generally capable of germination and fruiting. "spore-forming" or "spore-forming" bacteria are those that contain genes and other necessary abilities to produce spores under appropriate environmental conditions.

As used herein, a "combination" of two or more bacteria includes the physical co-presence of the two bacteria in the same material or product or in physically linked products, as well as the temporary co-administration or co-localization of the two bacteria.

As used herein, "subject" refers to any animal subject, including humans, laboratory animals (e.g., primates, rats, mice), livestock (e.g., dairy calves, sheep, goats, pigs, turkeys, chickens), and household pets (e.g., dogs, cats, rodents, etc.). The subject or patient may be healthy, or may have an infection caused by a gastrointestinal pathogen, or may be at risk for the infection caused by a gastrointestinal pathogen to develop or spread to others.

As used herein, "Shannon diversity index" refers to a diversity index that uses the following formula to illustrate the abundance and uniformity of species in a given population

Figure BDA0002304268610000171

Wherein H is the Shannon diversity index, R is the total number of species in the community, piIs the ratio of R consisting of the ith species. Higher values indicate a population with strong diversity and uniform distribution, and a value of 0 indicates only one species in a given population. Reference may further be made to the mathematical theory of communications by Shannon and Weaver, 1949, university of illinois publisher, ibaner, p 117.

As used herein, "antibiotic" refers to a substance that treats and/or prevents a bacterial infection by killing the bacteria, inhibiting the growth of the bacteria, or reducing the viability of the bacteria.

As used herein, an "intermittent dosing regimen" refers to administration of a therapeutic composition for a period of time (a treatment period) followed by a period of time during which treatment with the therapeutic composition is discontinued (a rest period). An intermittent dosing regimen may be expressed as a treatment period of days or weeks/a rest period of days or weeks. For example, an 4/1 intermittent dosing regimen refers to a treatment period of four weeks/day and a rest period of one week/day.

As used herein, a "continuous dosing regimen" refers to a dosing regimen in which the therapeutic composition is administered during a treatment period without a rest period. The therapeutic composition may be administered, for example, weekly, daily, or every other day or every third day throughout the treatment period of a continuous dosing regimen. Within a day of administration of the therapeutic composition, it may be administered in a single dose, or in multiple doses throughout the day.

As used herein, "dosing frequency" refers to the frequency of administration of a therapeutic composition over a given time period. The frequency of administration may be expressed as the number of administrations per given period of time, e.g., once per day, once per week or once every two weeks.

As used herein, "dosing interval" refers to the time elapsed between administration of multiple doses to a subject.

PD is a degenerative disease of the central nervous system, also known as idiopathic parkinsonism, or idiopathic parkinsonism. The cause of this disease is still unclear, but it is manifested as damage to the substantia nigra neurons of the brain, resulting in a deficiency of dopamine, a neurotransmitter. A reduction in dopamine levels can lead to motor impairment through irregular nerve discharges in the brain. The major dyskinesias associated with PD include tremor, rigidity, bradykinesia, and postural instability.

Current pharmacotherapy for parkinson's disease primarily employs dopaminergic drugs that either mimic dopamine or increase dopamine levels in the body. The most common therapy is levodopa, which is a metabolic precursor of dopamine. However, long-term levodopa treatment is often accompanied by dyskinesias, i.e. sudden, involuntary movements. In addition, long-term levodopa treatment can produce an "on-off" effect in which the patient normally moves "on" and reverts to PD-related dyskinesia "off". These side effects increase as levodopa treatment progresses, and the "off" period lasts longer. Other dopaminergic agents, anticholinergics and/or amantadine may also be used alone or in combination with levodopa therapy.

Surgical procedures, such as deep brain stimulation, have also been used to alleviate the symptoms of parkinson's disease. Recent studies have shown that certain regions of the brain are extremely active in PD, probably responsible for PD tremor, and that improvement of symptoms in mouse models and human PD by cortical stimulation has been proposed.

Many chronic diseases and gastrointestinal diseases have chronic infections/attacks as their underlying pathological conditions (e.g., PD). In one aspect, the present disclosure includes and relates to the use of fecal microbiota, one or more microbial species from fecal microbiota, active fragments or components from fecal microbiota for the treatment and/or prevention of various disease states (e.g. PD) associated with the presence of 'abnormal' microbiota in the gastrointestinal tract. The active fragment of a bacterium can be any active molecule isolated from such a bacterium by any known method of preparing/recognizing a bacterial active fragment and a protein secreted by the bacterium. These methods include, but are not limited to, the following: sonication, osmotic shock, detergent lysis, high pressure or transfer of suitable DNA to other organisms, such as bacteria, plants or animals, which are then used as feed additives as described previously. In one aspect, the active fragment or component of the bacterium is selected from the group consisting of a mycolate or derivative thereof, a polysaccharide, a lipopolysaccharide, a small peptide, a thiopeptide, a protein, a nucleic acid molecule, a metabolite, a cell wall component, or any combination thereof. In one aspect, the active fragment is a protein or a secretion. In another aspect, the active fragment is a secreted protein.

In one aspect, the present disclosure provides a method of treating PD in a subject in need of treatment, wherein the method comprises administering to the subject a pharmaceutically active dose of a therapeutic composition comprising live non-pathogenic fecal bacteria and/or sterile fecal filtrate. In another aspect, the present disclosure provides the use of a composition comprising live non-pathogenic fecal bacteria and/or a sterile fecal filtrate in the manufacture of a medicament for the treatment of PD.

In one aspect, the therapeutic composition comprises an isolated or purified population of live, non-pathogenic fecal bacteria. In one aspect, the therapeutic composition comprises an unselected fecal microbiota. In another aspect, the therapeutic composition comprises an unselected and substantially intact fecal microbiota. In another aspect, the therapeutic composition comprises a full spectrum fecal microbiota. In one aspect, a method comprises, in addition to a fecal microbiome-based treatment, simultaneously or sequentially, symptomatic anti-parkinson's disease treatment of a patient. In another aspect, in addition to fecal microbiome based treatment, a method further comprises administering to the patient bromocriptine, benztropine, levodopa, ropinirole, pramipexole, rotigotine, cabergoline, entacapone, tolcapone, amantadine, selegiline, or a combination thereof, either simultaneously or sequentially.

In one aspect, the fecal microbiome based PD therapy described herein is used in combination, sequentially, simultaneously or sequentially with another therapy selected from levodopa (with or without carbidopa), dopamine agonists, dopamine inactivating enzyme inhibitors (MAOB inhibitors and COMT inhibitors), anticholinergics, amantadine, and estrogens. In another aspect, the fecal microbiome based therapy described herein is used in combination with one or more drugs selected from levodopa, monoamine oxidase b (maob) inhibitors (e.g., selegiline and rasagiline), dopamine agonists (e.g., bromocriptine, pramipexole, ropinirole, transdermal rotigotine and injectable apomorphine), catechol-o-methyltransferase (COMT) inhibitors (e.g., tolcapone and entacapone), anticholinergic drugs (e.g., trihexyphenidyl, benztropine, octofenadrine, propidin and biperiden), amantadine.

In one aspect, the present disclosure provides a method of eliminating or reducing one or more symptoms of PD selected from tremor, slowed movement (bradykinesia), muscle stiffness, impaired posture and balance, loss of spontaneous movement, changes in speech, and changes in writing. In another aspect, the present disclosure provides a method of eliminating, ameliorating, alleviating, or reducing one or more symptoms of early PD. In another aspect, the treatment methods provided herein can reduce, alleviate, or control one or more symptoms of non-motor PD, such as depression, sleep disorders, dementia, psychosis, and hallucinations.

In another aspect, the treatment methods provided herein can reduce, alleviate or control one or more major motor symptoms of PD selected from tremor, bradykinesia, rigidity, and postural instability. In another aspect, the treatment methods provided herein can reduce, alleviate or control one or more secondary motor symptoms of PD selected from the group consisting of loss of gait perception, undersized font (handwriting contraction), facial expression (facial expression may appear less than usual), undesirable acceleration, postural kyphosis (with a tendency to anteversion), dystonia, impaired fine motor flexibility and motor coordination, impaired gross motor coordination, poor motor (reduced arm swing), akathisia, speech problems (such as soft voice or slurred speech due to insufficient muscle control), dysphagia, sexual dysfunction, cramps, watery lips, and excessive saliva due to reduced swallowing. In another aspect, the treatment methods provided herein can reduce, ameliorate or control one or more conditions in a PD patient selected from the group consisting of:

loss of smell and constipation

Rapid eye movement behavior disorder (sleep disorder)

Emotional disorders

Orthostatic hypotension (hypotension while standing)

Sleep disorders

Constipation

Bladder problems

Sexual problem

Excessive saliva

Weight loss or gain

Problems with vision and teeth

Fatigue and weakness

Depression (D)

Fear and anxiety

Skin problems

Cognitive problems such as memory difficulties, thought retardation, confusion and in some cases dementia, and

drug side effects, such as impulsivity.

In one aspect, the PD treatment described herein does not include or substantially does not include one or more side effects selected from nausea, somnolence, dizziness, headache, vomiting, dry mouth, blurred vision, constipation, difficulty emptying the bladder, difficulty sweating and increased heart rate, hypotension after standing, confusion, hallucinations, delusions, agitation, swelling of the lower legs and feet, swelling of the ankles, dyskinesia, orange urine, and psychosis.

In one aspect, the treated patient receives a pretreatment of oral broad spectrum antibacterial therapy for 7-10 days. In one aspect, a single-dose antibiotic regimen is used. In another aspect, a multi-dose antibiotic regimen is used. Exemplary antibiotic regimens include the following: (a) ciprofloxacin and metronidazole; (b) trimethoprim-sulfamethoxazole and metronidazole; (c) moxifloxacin; (d) amoxicillin/clavulanic acid. Other exemplary antibiotics include piperacillin/tazobactam, ampicillin/sulbactam, ticarcillin/clavulanic acid, imipenem, meropenem, tigecycline (when severe penicillin allergy). In another aspect, multiple drug regimens may consist of metronidazole and a third generation cephalosporin or fluoroquinolone, for example: ceftriaxone, cefotaxime, ciprofloxacin and levofloxacin.

In one aspect, the present disclosure provides a method of treating PD in a subject in need of treatment, wherein the method comprises administering to the subject a pharmaceutically active dose of a therapeutic composition comprising a live, non-pathogenic bacterium. In one aspect, the present disclosure provides a method of treating PD in a subject in need of treatment, wherein the method comprises administering to the subject a pharmaceutically active dose of a therapeutic composition comprising live, non-pathogenic fecal bacteria daily or weekly. In one aspect, the therapeutic composition is administered to a PD patient in need of treatment at least once daily or at least once weekly for at least two consecutive days or weeks. In one aspect, the therapeutic composition is administered at least once daily or at least once weekly for at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 consecutive days or weeks. In another aspect, the therapeutic composition is administered at least once daily or at least once weekly for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 consecutive weeks. In one aspect, the therapeutic composition is administered at least once daily or at least once weekly for up to 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 consecutive days or weeks. In another aspect, the therapeutic composition is administered at least once daily or at least once weekly for up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 consecutive weeks or months. In another aspect, the therapeutic composition is administered chronically at least once for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 consecutive months or years throughout the life cycle or indefinite period of the subject.

In one aspect, the therapeutic composition is administered to a PD patient in need of treatment at least twice daily or at least twice weekly for at least two consecutive days or weeks. In one aspect, the therapeutic composition is administered at least twice daily or at least twice weekly for at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 consecutive days or weeks. In another aspect, the therapeutic composition is administered at least twice daily or at least twice weekly for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 consecutive weeks. In one aspect, the therapeutic composition is administered at least twice daily or at least twice weekly for up to 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 consecutive days or weeks. In another aspect, the therapeutic composition is administered at least twice daily or at least twice weekly for up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 consecutive weeks or months. In another aspect, the therapeutic composition is administered chronically at least twice for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 consecutive months or years throughout the life cycle or indefinite period of the subject.

In one aspect, the therapeutic composition is administered to a PD patient in need of treatment at least three times daily or at least three times weekly for at least two consecutive days or weeks. In one aspect, the therapeutic composition is administered at least three times daily or at least three times weekly for at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 consecutive days or weeks. In another aspect, the therapeutic composition is administered at least three times daily or at least three times weekly for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 consecutive weeks. In one aspect, the therapeutic composition is administered at least three times daily or at least three times weekly for up to 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 consecutive days or weeks. In another aspect, the therapeutic composition is administered at least three times daily or at least three times weekly for up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 consecutive weeks or months. In another aspect, the therapeutic composition is chronically administered at least three times for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 consecutive months or years throughout the life cycle or indefinite period of the subject.

In one aspect, the present disclosure provides a method of treating PD in a subject in need of treatment, wherein the method comprises orally administering to the subject a pharmaceutically active dose of a therapeutic composition comprising a mixture of live non-pathogenic synthetic bacteria or a live non-pathogenic purified or extracted fecal microbiota, wherein the dose is administered on a dosing schedule of at least once or twice daily or at least once or twice weekly for at least three consecutive days or weeks. In another aspect, the dose is administered at least once, twice or three times daily, or at least once, twice or three times daily for 1 to 12 weeks, 2 to 12 weeks, 3 to 12 weeks, 4 to 12 weeks, 5 to 12 weeks, 6 to 12 weeks, 7 to 12 weeks, 8 to 12 weeks, 9 to 12 weeks, 10 to 12 weeks, 1 to 2 weeks, 2 to 3 weeks, 3 to 4 weeks, 4 to 5 weeks, 5 to 6 weeks, 6 to 7 weeks, 7 to 8 weeks, 8 to 9 weeks, 9 to 10 weeks, or 10 to 11 weeks.

In one aspect, the present disclosure provides a method of treating PD in a subject in need of treatment, wherein the method comprises a first dosing regimen followed by a second dosing regimen. In one aspect, the first dosing regimen comprises a therapeutic or induction dose. In one aspect, the first dosing regimen comprises a continuous dosing regimen. In another aspect, the second dosing regimen comprises a maintenance dose that is less than or equal to the pharmaceutically active dose of the first dosing regimen. In another aspect, the second dosing regimen lasts at least about 2, 4, 6, 8, 10, 12, 18, 24, 36, 48, 72, or 96 months. In one aspect, the second dosing regimen is continued throughout the life cycle or indefinitely for the treatment of the subject. In one aspect, the second dosing regimen is a continuous dosing regimen. In another aspect, the second dosing regimen is an intermittent dosing regimen. In another aspect, the second dosing regimen is an intermittent dosing regimen comprising a treatment period of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days followed by a rest period of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days. In another aspect, the second dosing regimen comprises administering the second dose (e.g., a maintenance dose) every other day, every third day, or every third, fourth, fifth, sixth, seventh, eighth, tenth, etc. day. In another aspect, the maintenance dose is administered chronically with or without titration (or otherwise altering the dose or dosing regimen). In one aspect, the interval between the first and second dosing regimens is at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 weeks. In another aspect, the second dosing regimen (e.g., maintenance dose) comprises a dose that is about 2, 5, 10, 50, 100, 200, 400, 800, 1000, 5000-fold or more lower than the dose used in the first dosing regimen (e.g., initial therapeutic dose). In another aspect, the second dosing regimen (e.g., maintenance dosing regimen) has the same or lower dosing frequency as the first dosing regimen (e.g., initial therapeutic dosing regimen). In another aspect, the second dosing regimen (e.g., maintenance dosing regimen) has a higher dosing interval than the first dosing regimen (e.g., initial therapeutic dosing regimen).

In one aspect, the first or second dosing regimen used in the method can be once per week, twice per week, or three times per week. The term "once weekly" refers to a once weekly administration, preferably on the same day of the week. By "twice weekly" is meant twice weekly administration, preferably on the same two days per week. By "three times per week" is meant three times a week administration, preferably on the same three days per week.

In one aspect, the subject receiving treatment is a subject already having PD. The disclosed therapeutic compositions can also prevent the onset of clinical symptoms of PD by administering to a clinically asymptomatic human subject who is genetically predisposed or susceptible to PD. A human subject that is genetically susceptible or predisposed to PD may be a human subject having a relative or relative exhibiting or having PD. In another aspect, the subject receiving treatment is a subject who is to be prevented from PD. In another aspect, the subject receiving treatment is susceptible or predisposed to PD. In another aspect, the subject receiving treatment is a subject diagnosed with PD. In one aspect, the subject receiving treatment is a patient in need of PD treatment. In another aspect, the immune function of the patient being treated is impaired. In another aspect, the patient receiving treatment is concurrently suffering from PD and gastrointestinal disease (e.g., chronic constipation, IBD, IBS, or clostridium difficile infection).

In one aspect, the subject receiving treatment has stage I, II, III, IV or V PD. In one aspect, the methods described herein are capable of improving the condition of a PD patient such that the patient's PD is re-staged for at least one or two stages (e.g., from stage III to stage II or from stage III to stage I). In another aspect, the methods described herein can prevent the deterioration of a PD patient, leaving the patient's PD stage unchanged or deteriorating more slowly. In another aspect, the methods described herein are capable of reducing the rate of progression of PD in a subject. In another aspect, the methods described herein are capable of delaying the need for symptomatic anti-parkinson's disease treatment in a subject. In one aspect, the patient is treated as described herein after being diagnosed with one or more early symptoms of parkinson's disease.

In one aspect, the subject receiving treatment is a human patient. In one aspect, the patient is a male patient. In one aspect, the patient is a female patient. In one aspect, the patient is a premature infant. In one aspect, the patient is a full term neonate. In one aspect, the patient is a neonate for less than four weeks. In one aspect, the patient is an infant. In one aspect, the patient is a toddler. In one aspect, the patient is a young child. In one aspect, the patient is a child. In one aspect, the patient is a juvenile. In one aspect, the patient is a pediatric patient. In one aspect, the patient is an elderly patient. In one aspect, the human patient is a pediatric patient under the age of about 18, 15, 12,10, 8, 6, 4,3, 2, or 1. In another aspect, the human patient is an adult patient. In another aspect, the human patient is an elderly patient. In another aspect, the human patient is a patient about 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, or 95 years old or older. In another aspect, the patient is about 1-5 years old, 2-10 years old, 3-18 years old, 21-50 years old, 21-40 years old, 21-30 years old, 50-90 years old, 60-90 years old, 70-90 years old, 60-80 years old, or 65-75 years old. In one aspect, the patient is a younger elderly patient (65-74 years old). In one aspect, the patient is an elderly patient of moderate age (75-84 years old). In one aspect, the patient is an elderly patient (>85 years old).

In one aspect, a method comprises administering the therapeutic composition orally, via an enema, or via a rectal suppository. In one aspect, the therapeutic compositions administered herein are formulated as enteric (and/or acid resistant) capsules or microcapsules, or as part of or administered with a food product, food additive, dairy product, soy product or derivative thereof, jelly, flavored liquid, ice cream, or yogurt. In another aspect, the therapeutic compositions administered herein are formulated as acid resistant enteric capsules. A therapeutic composition can be made into powder and sold together with food or beverage. The food or beverage may be a dairy product or a soy product. In another aspect, the food or food supplement comprises enteric and/or acid resistant microcapsules containing the therapeutic composition.

In one aspect, the therapeutic composition comprises a liquid culture. In another aspect, the therapeutic composition is lyophilized, pulverized, and powdered. And then can be injected, dissolved in, for example, saline, as an enema. Alternatively, the powder may be packaged into enteric and/or acid resistant capsules for oral administration. These capsules may take the form of enteric and/or acid resistant microcapsules. The powder may preferably be provided in a palatable form, as an additional ingredient to a beverage or as an additional ingredient to a food additive. In another aspect, the food product is yogurt. In one aspect, the powder may be reconstituted for injection by nasoduodenal perfusion.

In another aspect, the therapeutic compositions administered herein are in liquid, frozen, freeze-dried, spray-dried, foam-dried, lyophilized, or powder form. In another aspect, the therapeutic compositions administered herein are formulated for delayed or gradual enteric release. In another aspect, the therapeutic compositions administered herein comprise an excipient, saline, buffer, or fluid-glucose-cellobiose agar (RGCA) medium. In another aspect, the therapeutic compositions administered herein comprise a cryoprotectant. In one aspect, the cryoprotectant comprises polyethylene glycol, skim milk, erythritol, arabitol, sorbitol, glucose, fructose, alanine, glycine, proline, sucrose, lactose, ribose, trehalose, dimethyl sulfoxide (DMSO), glycerol, or a combination thereof.

In one aspect, the therapeutic compositions administered herein further comprise an acid inhibitor, an antacid, an H2 antagonist, a proton pump inhibitor, or a combination thereof. In one aspect, the therapeutic compositions administered herein are substantially free of non-biological material. In another aspect, the therapeutic compositions administered herein are substantially free of decellularized material selected from the group consisting of residual fiber, DNA, viral coat material, and inactive material.

In one aspect, the therapeutic composition further comprises or is supplemented with a prebiotic nutrient selected from a polyol, Fructooligosaccharide (FOS), fructooligosaccharide, inulin, Galactooligosaccharide (GOS), Xylooligosaccharide (XOS), polydextrose, monosaccharide, tagatose and/or mannooligosaccharide.

In one aspect, a method further comprises pre-treating the subject with an antibiotic composition prior to administering the therapeutic bacterial or microbiota composition. In one aspect, the antibiotic composition administered herein comprises an antibiotic selected from the group consisting of rifabutin, clarithromycin, clofazimine, vancomycin, rifampin, nitroimidazole, chloramphenicol, and combinations thereof. In another aspect, the antibiotic composition administered herein comprises an antibiotic selected from the group consisting of rifaximin, rifamycin derivatives, rifampin, rifabutin, rifapentine, rifalazil, bicyclomycin, aminoglycosides, gentamicin, neomycin, streptomycin, paromomycin, vancomycin, mutamycin, sisomicin, netilmicin, ritemicin, kanamycin, aztreonam macrolide, clarithromycin, dirithromycin, roxithromycin, telithromycin, azithromycin, bismuth subsalicylate, vancomycin, streptomycin, fidaxomycin, amikacin, arbekacin, neomycin, netilmicin, paromomycin, erythromycin, tobramycin, apramycin, and combinations thereof. In another aspect, a method further comprises pre-treating the subject with an anti-inflammatory drug prior to administering the therapeutic bacterial or microbiota composition.

In one aspect, a method achieves a rate of remission, cure, response, or regression of PD of at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, or 99%. In one aspect, the UPDRS score decreases by more than 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 after 4, 8, or 12 weeks of treatment with one treatment method. In another aspect, the UPDRS score decreases by more than 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 after 4, 8, or 12 weeks of treatment with one treatment method in at least 10%, 20%, 30%, 50%, 60%, 70%, 80%, or 90% of the patients in the patient population. In one aspect, the UPDRS score is reduced by at least 10%, 20%, 30%, 50%, 60%, 70%, 80%, or 90% after 4, 8, or 12 weeks of treatment with one treatment method as compared to baseline (e.g., immediately prior to treatment). In one aspect, the treatment method achieves at least a 10%, 20%, 30%, 50%, 60%, 70%, 80%, or 90% reduction in UPDRS score in at least 10%, 20%, 30%, 50%, 60%, 70%, 80%, or 90% of patients after 4, 8, or 12 weeks of treatment as compared to baseline (e.g., immediately prior to treatment). Although UPDRS is used as one example of characterizing and quantifying PD severity, a similar or comparable system (e.g., MDS-UPDRS) may also be used. In each of the examples in which the UPDRS is mentioned in this application, another similar scale or system may be used in place of the UPDRS. Such replacement systems include, for example, the UPDRS Disability Score (UDS) (Mart Itez-Martini et al, Neurologia.2000, 11 months; 15(9): 382-7). In another aspect, a decrease in the UPDRS score referred to herein is a decrease in the UPDRS total score. In another aspect, a decrease in the UPDRS score as referred to herein refers to a decrease in the score of any one or both of (1) mental, behavioral, and emotional, (2) Activities of Daily Living (ADL), and (3) motor components.

In one aspect, the treatment methods described herein enable rapid improvement in PD symptoms. In another aspect, the patient's PD symptoms improve after five or fewer fecal microbiome infusions by colonoscopic delivery or enema for one week. In another aspect, a PD patient improves symptoms after one course of treatment as described herein in the absence of treatment for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 weeks.

In another aspect, a PD patient is treated with fecal microbe-based therapy, including fecal microbe fortificationThe induction phase of the drug is followed by the maintenance phase of the weak booster dose. In one aspect, the induction phase comprises injection of fresh fecal microbiome by colonoscopic delivery or enema. In one aspect, the induction phase comprises 5 to 10 injections of fresh fecal microbiome by colonoscopic delivery or enema over 1 to 2 weeks. In another stage, the induction stage comprises the ingestion of acid resistant/slow release capsules containing lyophilized unselected donor fecal microbiota. In one aspect, the maintenance phase comprises ingestion of a capsule containing a lyophilized unselected donor fecal microbiota. In another aspect, the maintenance phase comprises ingestion of 6 or more, 4 or more, or 2 or more capsules per day, wherein each capsule comprises about 1010To 1013Total number of individual cells. In another aspect, the maintenance phase comprises ingestion of 6 or more, 4 or more, or 2 or more capsules every other day, wherein each capsule contains about 1010To 1013Total number of individual cells.

In one aspect, the pharmaceutically active dose is contained per about 200mg of the pharmaceutical composition. In one aspect, the pharmaceutically active dose is contained per about 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 450, 500, 750, 1000, 1500, or 2000mg of the pharmaceutical composition.

In one aspect, a pharmaceutically active or therapeutically effective dose comprises at least about 105、106、107、108、109、1010、1011、1012、1013、1014Or 1015cfu. In another aspect, the pharmaceutically active therapeutically effective dose comprises up to about 105、106、107、108、109、1010、1011、1012、1013、1014Or 1015cfu. In another aspect, the pharmaceutically active therapeutically effective dose is selected from 108To 1014cfu,109To 1013cfu,1010To 1012cfu,109To 1014cfu,109To 1012cfu,109To 1011cfu,109To 1010cfu,1010To 1014cfu,1010To 1013cfu,1011To 1014cfu,1011To 1013cfu,1012To 1014cfu, and 1013To 1014cfu. In one aspect, the pharmaceutical composition comprises a unit weight of about 0.2, 0.4, 0.6, 0.8 or 1.0 grams or a unit volume of about 0.2, 0.4, 0.6, 0.8 or 1.0 milliliters of the aforementioned pharmaceutically active or therapeutically effective dose.

In one aspect, a pharmaceutically active or therapeutically effective dose comprises at least about 105、106、107、108、109、1010、1011、1012、1013、1014Or 1015Individual cells or spores. In another aspect, the pharmaceutically active or therapeutically effective dose comprises up to about 105、106、107、108、109、1010、1011、1012、1013、1014Or 1015A total of several cells or spores. In another aspect, the pharmaceutically active or therapeutically effective dose is selected from 108To 1014,109To 1013,1010To 1012,109To 1014,109To 1012,109To 1011,109To 1010,1010To 1014,1010To 1013,1011To 1014,1011To 1013,1012To 1014And 10 are13To 1014Individual cells or spores. In one aspect, the pharmaceutically active or therapeutically effective dose of cell counts is directed to living cells. In one aspect, the pharmaceutical composition comprises a unit weight of about 0.2, 0.4, 0.6, 0.8 or 1.0 grams or a unit volume of about 0.2, 0.4, 0.6, 0.8 or 1.0 milliliters of the aforementioned pharmaceutically active or therapeutically effective dose. In one aspect, a pharmaceutically active or therapeutically effective dose comprises 1010To 1012And (4) cells. In another aspect, a pharmaceutically active or therapeutically effective dose comprises each capsule 1010To 1012And (4) cells.

In one aspect, the therapeutic compositions administered herein comprise fecal bacteria. In one aspect, the therapeutic compositions administered herein comprise one or more, two or more, three or more, four or more or five or more isolated, purified or cultured microorganisms selected from the group consisting of clostridium, bacillus, coriolus, bacteroides, eubacterium, clostridium, propionibacterium, lactobacillus, ruminococcus, escherichia, gemfibrobacter, desulfomonas, peptostreptococcus, bifidobacterium, coprococcus, doleria and candida.

In one aspect, the therapeutic composition administered herein comprises at least one, at least two, at least three, at least four, at least five, at least six, or at least seven fecal microorganisms selected from: common species of Bacteroides fragilis, Coprinus aerogenes, Bacteroides fragilis, Streptococcus digestus type II, Parabacteroides dieldii, Clostridium prasukii, enterococcus faecalis, Coprinus aerogenes type III, Streptococcus digestus type I, Ruminococcus ruminis, Bifidobacterium adolescentis, Blastomyces formate, Bifidobacterium longum, Eubacterium indolens, Pediococcus stractici, Eubacterium recta, Shigella exneri, Clostridium mollis, Bacteroides fragilis type A, Eubacterium bipinnata, Bifidobacterium infantis, Eubacterium procum III-F, Pediobolus, Pseudolavonobacter asiaticus, Ruminococcus albus, Doller longum, Eubacterium hophragmarius, Eubacterium ventricoides type I, Fusobaterirussia, Ruxococcus ovatus, Runella proctolicus, Clostridium ramosum, Lactobacillus lisi, Ruminococcus fasciatus, Vibrio parahaemolyticus amino acid fermentation, Vibrio, species of Bacteroides fragilis, Lactobacillus gassericatarrhalis, Streptococcus fragilis species, Bacteroides AR, Peptococcus dextrinus, Aerositeshhrus, Eubacterium cylindricum, Eubacterium ruminants, Eubacterium CH-1, Staphylococcus epidermidis, Streptococcus digestus BL, Eubacterium mucosae, Yersinia extremophila, Bacteroides L, Clostridium mortiferum type I, Clostridium navigatum, Clostridium innocuous, Clostridium ramosum, Propionibacterium acnes, Ruminococcus flavus, Ruminococcus AT, Peptococcus AU-1, Vibrio fragilis oval species, Bacteroides L-1 and L-5, Fusobacterium nucleatum, Clostridium mortiferum, Escherichia coli, measles, Proteus macrofengolensis, Peptococcus G, -AU-2, Streptococcus intermedius, Ruminococcus acidophilus CO, Gibberella X, Peptococcus BH, -CC, Eubacterium gracilis, Eubacterium ramosum, Clostridium clostridia species, Bacteroides coagulans, Prevotella oral cavity, Prevotella, Peptococcus neoides acidium japonicum, Peptococcus, Prevotella ruminis, Odoribacter splanchnicus, Desuifonas pigra, Lactobacillus G, Vibrio succinate A and combinations thereof.

In one aspect, the therapeutic composition administered herein does not comprise live bacteroides, clostridia, propionibacteria, lactobacilli, ruminococcus, escherichia, gemfibrobacter, desulfomonas, peptostreptococcus, bifidobacterium, candida, or any combination thereof. In another aspect, the therapeutic compositions administered herein do not comprise viable Bacteroides fragilis common species, Coprinus aerogenes, Bacteroides fragilis polymorphic species, Pediococcus neoformans type II, Parabacteriodes diesei, Clostridium praecox, enterococcus faecalis, Coprinus aerogenes type III, Pediococcus neoformans type I, Ruminococcus ruminis, Bifidobacterium adolescentis, Blastomyces formis, Bifidobacterium longum, Eubacterium inerticus, Pediococcus contortus, Eubacterium recta, Eubacterium pickeri, Bacteroides exubertii, Clostridium molesta, Eubacterium fragilis type A, Eubacterium biformis, Bifidobacterium infantis, Eubacterium procumbens III-F, Pediococcus chaperone, Pseudolavonofrer calf bacteria strain, Runellus albus, Dollerella longum, Eubacterium johnsonii, Eubacterium ventricosum, Fusobacterium ruscus, Eubacterium procumbellatum, Clostridium ramosum, Lactobacillus reuteri, Vibrio rei, Vibrio fural, Clostridium butyricum, Clostridium perfringens, Clostridium bifidum type I, Clostridium difficile, fermented aminoacid coccus, Eubacterium ventriosum, Bacteroides fragilis species, Bacteroides AR, Pediococcus dextrinus, Aerostipes hadrus, Eubacterium cylindricum, Eubacterium ruminants, Eubacterium CH-1, Staphylococcus epidermidis, Streptococcus digestus BL, Eubacterium mucosus, Yersinia stipitis, Bacteroides L, Clostridium mortiferum type I, Clostridium navigatum, Clostridium innocuum, Clostridium ramosum, Propionibacterium acnes, Ruminococcus flavus, Ruminococcus AT, Pediococcus AU-1, Pedioides oval species, Bacteroides L-1 and L-5, Clostridium nucleatum, Clostridium mortiferum, Escherichia coli, measles probiotic, Fagagord bacteria, Pediococcus G, -twin-2, Medococcus, Ruminococcus acidophilus, Ruminococcus CO, Gibberella X, enterococcus faecalis, -CC, Eubacterium gracilicate, Clostridium, Eubacterium ramosum, Clostridium clostridia, Bacteroides coagulans, Prevotella abortus, Prevotella ruminicola, Odoribacter splanchnicus, Desufomonas pigra, Lactobacillus G, Vibrio succinate A, and combinations thereof.

In one aspect, the therapeutic compositions administered herein comprise fecal microbiota. In another aspect, the preparation of fecal microbiota as used herein includes a treatment method selected from the group consisting of ethanol treatment, detergent treatment, heat treatment, radiation and sonication. In another aspect, the preparation of fecal microbiota as used herein does not involve a treatment method selected from the group consisting of ethanol treatment, detergent treatment, heat treatment, radiation and sonication. In one aspect, the preparation of a fecal microbiota as used herein involves a separation step selected from the group consisting of density gradient, filtration (e.g., screen, nylon mesh), and chromatography. In another aspect, the preparation of a fecal microbiota as used herein does not involve a separation step selected from the group consisting of density gradient, filtration (e.g., screen, nylon mesh), and chromatography. In another aspect, fecal microbiota as used herein includes the complete fecal microbiota of the donor. In another aspect, the therapeutic compositions administered herein comprise a fecal microbiota that is substantially free of eukaryotic cells from a fecal microbiota donor.

In another aspect, the therapeutic compositions administered herein comprise a fecal microbiota supplemented, enhanced or enhanced with fecal microbes. In one aspect, the fecal microbiota is supplemented with non-pathogenic (or reduced pathogenic) bacteria of the genera clostridium, corinth, dorsalmonella, ruminococcus, coprococcus, prevotella, veyonococcus, bacteroides, bacillus, or combinations thereof. In another aspect, the therapeutic compositions administered herein comprise a fecal microbiota supplemented, augmented or enhanced with veillonella, firmicutes, proteobacteria, bacteroides, or a combination thereof. In another aspect, the therapeutic compositions administered herein comprise a fecal microbiota supplemented with fecal bacterial spores. In one aspect, the fecal bacterial spore is a clostridial spore, a bacillus spore, or both.

In one aspect, the therapeutic composition comprises a fecal microbiota from a subject selected from a human, a cow, a dairy calf, a ruminant, a sheep, a goat, or a deer. In another aspect, the therapeutic composition may be administered to a subject selected from a human, a cow, a dairy calf, a ruminant, a sheep, a goat or a deer. In one aspect, the therapeutic composition is substantially or almost free of odor.

In one aspect, the therapeutic compositions provided or administered herein comprise a fecal microbiota having a shannon diversity index greater than or equal to 0.3, greater than or equal to 0.4, greater than or equal to 0.5, greater than or equal to 0.6, greater than or equal to 0.7, greater than or equal to 0.8, greater than or equal to 0.9, greater than or equal to 1.0, greater than or equal to 1.1, greater than or equal to 1.2, greater than or equal to 1.3, greater than or equal to 1.4, greater than or equal to 1.5, greater than or equal to 1.6, greater than or equal to 1.7, greater than or equal to 1.8, greater than or equal to 1.9, greater than or equal to 2.0, greater than or equal to 2.1, greater than or equal to 2.2, greater than or equal to 2.3, greater than or equal to 2.4, greater than or equal to 2.5, greater than or equal to 3.0, greater than or equal to 3.1, greater than or equal to 3.2, greater than 3.3, greater than or equal to 3.4, greater than or equal to 3.5, greater than 3.8, greater than, Greater than or equal to 3.9, greater than or equal to 4.0, greater than or equal to 4.1, greater than or equal to 4.2, greater than or equal to 4.3, greater than or equal to 4.4, greater than or equal to 4.5, or greater than or equal to 5.0. In another aspect, the therapeutic composition comprises a fecal microbiota having a shannon diversity index of 0.1 to 3.0, 0.1 to 2.5, 0.1 to 2.4, 0.1 to 2.3, 0.1 to 2.2, 0.1 to 2.1, 0.1 to 2.0, 0.4 to 2.5, 0.4 to 3.0, 0.5 to 5.0, 0.7 to 5.0, 0.9 to 5.0, 1.1 to 5.0, 1.3 to 5.0, 1.5 to 5.0, 1.7 to 5.0, 1.9 to 5.0, 2.1 to 5.0, 2.3 to 5.0, 2.5 to 5.0, 2.7 to 5.0, 2.9 to 5.0, 3.1 to 5.0, 3.3 to 5.0, 3.5 to 5.0, 3.9 to 5.0, or 1.5 to 5.0. In one aspect, the shannon diversity index is calculated at the phylum level. On the other hand, the shannon diversity index is calculated at the family level. In one aspect, the shannon diversity index is calculated at genus level. On the other hand, the shannon diversity index is calculated at the species level. In another aspect, the therapeutic composition comprises a microbiota preparation that is similar in proportional content to the normal healthy human fecal microbiota.

In another aspect, the therapeutic composition comprises fecal bacteria from at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 different families. In one aspect, the therapeutic compositions provided or administered herein comprise a fecal microbiota comprising no greater than 0.05%, 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, or 10% by weight of non-biological/biological material. In another aspect, a therapeutic composition provided or administered herein comprises a fecal microbiota comprising no greater than 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% by weight of non-biological/biological material. In another aspect, the therapeutic compositions provided or administered herein comprise, consist of, or consist essentially of particles of non-biological material and/or particles of biological material of a fecal sample that passes through a sieve, column, or similar filtration device having a sieve, reject, or particle filter of a size of 2.0mm, 1.0mm, 0.33mm, 0.5mm, 0.25mm, 0.212mm, 0.180mm, 0.150mm, 0.125mm, 0.106mm, 0.090mm, 0.075mm, 0.063mm, 0.053mm, 0.045mm, 0.038mm, 0.032mm, 0.025mm, 0.020mm, 0.01mm, 0.002mm, 0.1mm, or 0.2 mm. "non-biological material" does not include excipients such as pharmaceutical non-active substances added to the treated fecal material, e.g., cryoprotectants. "biological material" refers to living material in feces, including microorganisms, and microorganisms including prokaryotic cells, such as bacteria and archaea (e.g., living prokaryotic cells and spores that can be encysted to become living prokaryotic cells); eukaryotic cells, such as protozoa, fungi, and viruses. In one aspect, "biological material" refers to living material, such as microorganisms, eukaryotic cells, and viruses, that are present in the colon of a normal, healthy human. In one aspect, the therapeutic compositions provided or administered herein comprise a human fecal extract, wherein the composition is substantially odorless. In one aspect, the therapeutic compositions provided or administered herein comprise a fecal material or fecal flora preparation in a lyophilized, crude, semi-purified, or purified preparation.

In one aspect, the fecal microbiota in the therapeutic composition comprises a highly refined or purified fecal flora, e.g., substantially free of non-flora fecal material. In one aspect, the fecal microbiota can be further processed, for example, by microfiltration before screening, after screening, or both before and after screening. In another aspect, the highly purified fecal microbiota product is ultrafiltered to remove macromolecules, but retains a therapeutic microbial flora, such as bacteria.

In another aspect, the fecal microbiota in the therapeutic compositions used herein comprises or consists essentially of a substantially isolated or purified fecal flora or an intact (or substantially intact) microbiota that is (or comprises) at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9% isolated or pure isolates of a fecal flora or has no more than about 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, or 1.0% or more of non-fecal flora material; alternatively, a substantially isolated, purified or substantially intact microbiota as described in WO2012/122478Al to Sadowsky et Al, or WO2012/016287a2 to Borody et Al.

In one aspect, the fecal microbiota in the therapeutic composition comprises substantially intact or unselected fecal microbiota, recombinant fecal material, or synthetic fecal material of the donor. In another aspect, the fecal microbiota in the therapeutic composition does not comprise an antibiotic-resistant population. In another aspect, the therapeutic composition comprises a fecal microbiota and is substantially free of foreign material (e.g., non-biological material, including decellularized material, such as residual fiber, DNA, RNA, viral coat material, non-active material; and biological material, such as eukaryotic cells from a fecal donor).

In one aspect, the fecal microbiota in the therapeutic compositions used herein is from fresh homologous feces from a disease screen or equivalent freeze-dried and recombinant feces. In one aspect, the fresh homologous feces do not include an antibiotic resistant population. In another aspect, the fecal microbiota in the therapeutic composition is from a synthetic fecal composition. In one aspect, the synthetic fecal composition comprises a preparation of active flora that is preferably similar in proportional amount to normal healthy human fecal flora that does not include antibiotic resistant populations. Suitable microorganisms may be selected from the following: bacteroides, eubacterium, Clostridium, Propionibacterium, Lactobacillus, Ruminococcus, Escherichia, Gibberella, Clostridium, Desulfuromonas, Peptostreptococcus, Bifidobacterium, Coriolus, enterococcus, Doller's and Ruminococcus.

In one aspect, the therapeutic composition for use in the treatment disclosed herein comprises a sterile stool filtrate or a non-cellular stool filtrate. In one aspect, the sterile fecal filtrate is derived from donor feces. In another aspect, the sterile fecal filtrate is derived from a cultured microorganism. In another aspect, the sterile fecal filtrate comprises non-cellular, non-particulate fecal constituents. In one aspect, the sterile fecal filtrate is prepared as described in WO2014/078911 published 5-month-30-year 2014. In another aspect, a sterile fecal filtrate is prepared as described by Ott et al in Gastroenterology152:799-911 (2017).

In one aspect, the stool filtrate contains secreted, excreted, or other liquid components or microbiota, such as bioactive molecules (BAMs), which may be antibiotics or anti-inflammatory agents, preserved, retained, or reconstituted in the flora extract.

In one aspect, an exemplary therapeutic composition includes starting material from a donor defining a donor pool, wherein the donor provides centrifuged stool, which is then subjected to very high levels of filtration using, for example, a metal screen or a microporous filter or equivalent, to ultimately allow only bacteria-derived cells to remain, e.g., typically less than about 5 microns in diameter. After initial centrifugation, the solid material is separated from the liquid, and then the solid is filtered in a gradually decreasing size filter and tangential filter, for example using microfiltration, and optionally also including filtration using nanomembranes. Filtration may also be accomplished by a sieve as described in WO2012/122478, but in contrast using a sieve of less than 0.0120mm, down to about 0.0110mm, will result in the eventual presence of only bacterial cells.

The supernatant separated during centrifugation is now removed and gradually filtered through, for example, microfiltration or equivalent systems, resulting in a liquid that is finely filtered through an about 0.22 micron filter. Thereby removing all particulate matter, including all biological matter, including bacteria and viruses. Sterile products are then obtained, but with the aim of removing the bacteria but retaining their secretions, in particular antibacterial bacteriocins, bacterially derived cytokine-like products and all the accompanying Biologically Active Molecules (BAMs), including: thuringiensis (which are secreted by bacilli in donor feces), bacteriocins (including colicin, troudulixine or pulainicine or microcin or subtilosin a), lantibiotic (including nisin, subtilisins, epidermins, mutanins, mecicins, actagardine, cinnamycin), nisin, and other antibacterial or anti-inflammatory compounds.

In one aspect, the therapeutic composition for use herein comprises a recombinant fecal flora consisting essentially of a combination of a purified fecal microbiota and an acellular fecal filtrate. In another aspect, the therapeutic composition used herein comprises a purified fecal microbiota supplemented with one or more non-cellular, non-particulate fecal constituents. In one aspect, the therapeutic compositions used herein comprise one or more non-cellular, non-particulate fecal constituents. In one aspect, the one or more non-cellular, non-particulate fecal components include synthetic molecules, biologically active molecules produced by fecal microorganisms, or both. In another aspect, the one or more non-cellular, non-particulate fecal constituents include bioactive proteins or peptides, micronutrients, fats, sugars, small carbohydrates, trace elements, mineral salts, ash, mucus, amino acids, nutrients, vitamins, minerals, or any combination thereof. In one aspect, the one or more non-cellular, non-particulate fecal component comprises one or more biologically active molecules selected from the group consisting of bacteriocins, lantibiotic, and nisin. In another aspect, the one or more acellular non-particulate fecal component comprises one or more bacteriocins selected from the group consisting of colicin, troudulixine, pulainicine, microcin, and subtilosin a. In one aspect, the one or more non-cellular, non-particulate fecal component comprises one or more lantibiotic selected from the group consisting of thuringiensis, nisin, subtilin, epidermin, mutanolysin, spinosad, actagardine, and cinnamycin. In another aspect, the one or more non-cellular, non-particulate fecal components includes an anti-spore compound, an antibacterial compound, an anti-inflammatory compound, or any combination thereof. In another aspect, the one or more non-cellular, non-particulate fecal components includes an interleukin, a cytokine, a leukotriene, an eicosanoid, or any combination thereof.

In another aspect, the treatment methods provided herein include the use of fecal bacterial cells, e.g., a partial or complete representation of the microbiota of the human gastrointestinal tract, and isolation, processing, filtration, concentration, reconstitution and/or artificial liquid components of the microbiota (e.g., fecal filtrate), including bacterial secretion products, e.g., bacteriocins (protein toxins produced by bacteria, including colicin, trouulixin or pulainicidine, or microcin or subtilisina), lantic (a class of peptide antibiotics, which contain the characteristic polycyclic thioether amino acids lanthionine or methyllanthionine, as well as the unsaturated amino acids dehydroalanine and 2-aminoisobutyric acid; including thuringicin (which is secreted by bacilli in donor feces), nisin, surfactin, epidermin, mutanolysin, mersacidin, cinnamycin, nisin (family of pore-forming peptide toxins) and other antibacterial or anti-inflammatory compounds and/or microbial populations derived from microbial populations Additional bioactive molecules (BAMs) produced by bacteria or other microorganisms, and/or found in the "liquid component" of a microbiota.

In one aspect, the fecal bacteria-based therapeutic composition is used concurrently with the fecal non-cell filtrate-based therapeutic composition. In another aspect, the patient is treated with the first fecal non-cell filtrate-based therapeutic composition prior to administering the second fecal bacteria-based therapeutic composition to the patient, or vice versa. In another aspect, the method of treatment comprises three steps: first, antibiotic pretreatment to unselectively remove infectious pathogens; second, a fecal non-cell filtrate based treatment step to further inhibit selected infectious agents; third, the patient is administered a fecal bacteria-based therapeutic composition to reconstitute the functional gut microbiome.

In one aspect, the therapeutic composition is combined with other adjuvants, such as antacids (e.g., gastric energy, gastric caine pastilles, gastric gel) to inhibit bacterial inactivation in the stomach. On the other hand, acid secretion in the stomach can also be pharmacologically inhibited with H2 antagonists or proton pump inhibitors. An example of an H2 antagonist is ranitidine. An example of a proton pump inhibitor is omeprazole. In one aspect, the acid inhibitor is administered prior to or in conjunction with the therapeutic composition.

In one aspect, the therapeutic composition is in the form of: enema compositions which can be reconstituted with a suitable diluent; enteric capsules; enteric microcapsules; acid resistant tablets; acid-resistant capsules; acid-resistant microcapsules; powders reconstituted with a suitable diluent by nasointestinal infusion or colonoscopic infusion; oral powders reconstituted with appropriate diluents, flavoring agents and gastric acid inhibitors; a powder for reconstitution with a food or beverage; or a food or food supplement comprising enteric and/or acid resistant microcapsules of a composition, powder, gel or liquid.

In one aspect, the method of treatment achieves a cure of PD, a reduction in symptoms, or a reduction in the percentage of symptoms. In another aspect, the method of treatment also affects changes in the flora. The change in flora is preferably as "near complete" as possible and the flora is replaced by living organisms which will expel any remaining original flora. Typically, the alteration of the intestinal flora comprises introducing a predetermined series of flora into the gastrointestinal system, and thus in a preferred form the method of treatment comprises substantially or completely replacing pathogenic intestinal flora in a patient in need of such treatment.

In another aspect, the therapeutic composition can be provided with a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" refers to a non-toxic solvent, dispersant, excipient, adjuvant, or other substance with which the viable bacteria are mixed, so as to form a pharmaceutical composition, such as a dosage form capable of being administered to a patient. The pharmaceutically acceptable carrier may be a diluent, adjuvant, excipient or acid-resistant capsule ingredient in liquid (e.g. saline), gel or solid form. Suitable diluents and excipients include pharmaceutical grade normal saline, dextrose, glycerol, mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, magnesium carbonate, and the like, and combinations thereof. In another aspect, the therapeutic composition may comprise auxiliary substances, such as wetting or emulsifying agents, stabilizing agents, or ph buffering agents. In one aspect, the therapeutic composition comprises about 1% -5%, 5% -10%, 10% -15%, 15% -20%, 20% -25%, 25% -30%, 30% -35%, 40% -45%, 50% -55%, 1% -95%, 2% -95%, 5% -95%, 10% -95%, 15% -95%, 20% -95%, 25% -95%, 30% -95%, 35% -95%, 40% -95%, 45% -95%, 50% -95%, 55% -95%, 60% -95%, 65% -95%, 70% -95%, 45% -95%, 80% -95%, or 85% -95% of the active ingredient. In one aspect, the therapeutic composition comprises about 2% -70%, 5% -60%, 10% -50%, 15% -40%, 20% -30%, 25% -60%, 30% -60%, or 35% -60% active ingredient.

In one aspect, the therapeutic composition can be incorporated into a tablet, infusion, pill, capsule, or premix. Such active ingredients may be formulated into such dosage forms by methods well known in the art of pharmaceutical formulation. See, for example, U.S. patent No. 4,394,377. Filling gelatin capsules with any desired form of the active ingredient makes it easy to produce capsules. If desired, these materials may be diluted with an inert powdered diluent, such as sugar, starch, powdered milk, purified crystalline cellulose, or the like, to increase the volume and facilitate filling of the capsules.

In one aspect, conventional formulation techniques can be used to prepare tablets containing the therapeutic composition. In addition to the active ingredient, tablets may contain binders, disintegrants, absorbents, binders, and lubricants. Typical bases include lactose, sugar, sodium chloride, starch and mannitol. Starch, like alginic acid, is also a good disintegrant. Surfactants such as sodium lauryl sulfate and sodium dioctylsulfosuccinate are also sometimes used. Commonly used absorbents include starch and lactose. Magnesium carbonate is also suitable for oily substances. The binder may use, for example, gelatin, gum, starch, dextrin, polyvinylpyrrolidone, and various cellulose derivatives. Common lubricants include magnesium stearate, talc, paraffin, various metal soaps, and polyethylene glycol.

In one aspect, for the preparation of solid compositions such as tablets, the active ingredient is mixed with a pharmaceutical carrier, for example, conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, or other pharmaceutical diluents such as water, to form a solid preformulation composition comprising a homogeneous mixture of the compositions of the present invention. When these preformulation compositions are described as homogeneous, it is meant that the active ingredient is dispersed uniformly throughout the entire composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. The solid preformulation compositions are then subdivided into unit dosage forms of the type described above containing the desired amount of active ingredient (e.g., at least about 10)5、106、107、108、109、1010、1011、1012Or 1013cfu or total cell number). The therapeutic compositions used herein can be flavored.

In one aspect, the therapeutic composition can be a tablet or pill. In one aspect, the tablets or pills may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, a tablet or pill can include an inner dosage and an outer dosage component, the latter being in the form of a film-coat over the former. The two components may be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release. Such enteric layers or coatings may be formed using a variety of materials, including various polymeric acids and mixtures of polymeric acids with shellac, cetyl alcohol and cellulose acetate, and the like.

In one aspect, the therapeutic composition can be an infusion. In one aspect, the infusion is prepared by selecting a saline suspension form of the therapeutic composition. By preparing a suspension of one ingredient in an aqueous solution with another ingredient, a water soluble form of one ingredient may be used in combination with a water insoluble form of the other ingredient. The water-insoluble form of either active ingredient may be prepared as a suspension or in some physiologically acceptable solvent such as polyethylene glycol. Suspensions of any one of the active ingredients in water-insoluble form can be prepared in oils such as peanut, corn, sesame oil, and the like; prepared in glycols such as propylene glycol or polyethylene glycol; or in water, depending on the solubility of the particular active ingredient. Suitable physiologically acceptable adjuvants may be required in order to keep the active ingredient suspended. Adjuvants may include and be selected from thickening agents such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and alginates. Surfactants are commonly used to suspend active ingredients, particularly the fat-soluble propionate enhancing compounds. Most suitable for preparing suspensions in liquid non-solvents are alkylphenol polyethylene oxide adducts, naphthalene sulfonates, alkylbenzene sulfonates and polyoxyethylene sorbitan esters. In addition, many substances that affect the hydrophilicity, density and surface tension of the liquid may in individual cases help to prepare the suspension. For example, silicone antifoams, glycols, sorbitol and sugars may be useful suspending agents.

In one aspect, a therapeutic composition comprises one or more, two or more, three or more, or four or more non-pathogenic spores of a Clostridium species selected from the group consisting of different Clostridium species, Clostridium argentifolium, Clostridium pasteurianum, Clostridium botulinum, Clostridium cadaveri, Clostridium botulinum, Clostridium cryptum, Clostridium shore, Clostridium clostridia, Clostridium sporum, Clostridium \35890Clostridium, Clostridium swineri, Clostridium fetchii, Clostridium goeri, Clostridium glycolurium, Clostridium hemolytic, Clostridium spear, Clostridium histolyticum, Clostridium indolens, Clostridium dyscrasum, Clostridium mucronatum, Clostridium novellum, Clostridium orotate, Clostridium paraputrefaciens, Clostridium perfringens, Clostridium putrescentium, Clostridium putrescentiale, Clostridium sartans, Clostridium flaccida, Clostridium scinum, Clostridium septicum, Clostridium sorangium, Clostridium difficile, Clostridium spirillum, Clostridium producens, Clostridium subterminale, Clostridium difficile, Clostridium perfringens, Clostridium difficile, Clostridium auto, Clostridium tetani, clostridium welchii and clostridium villosum.

In one aspect, a therapeutic composition comprises a purified, isolated, or cultured live non-pathogenic clostridium and a plurality of purified, isolated, or cultured live non-pathogenic microorganisms from one or more genera selected from the group consisting of corylinus, coprococcus, dorferia, eubacterium, and ruminococcus. In another aspect, a therapeutic composition comprises a plurality of purified, isolated, or cultured live non-pathogenic microorganisms from one or more genera selected from the group consisting of clostridium, chrysosporium, coprococcus, dorsalis, eubacterium, and ruminococcus.

In one aspect, a therapeutic composition comprises two or more genera selected from the genera chrysosporium, coprococcus, dorferia, eubacterium, and ruminococcus. In another aspect, a therapeutic composition comprises two or more genera selected from the genera coprococcus, dorsalomyces, eubacterium, and ruminococcus. In another aspect, a therapeutic composition comprises one or more, two or more, three or more, four or more, or five or more species selected from the group consisting of coprococcus dexterous, coprococcus, Dorea longicantina, shigella, eubacterium voluminoides, eubacterium hollisae, eubacterium proctosum, and streptococcus contortus.

In one aspect, a therapeutic composition comprises at least about 105、106、107、108、109、1010、1011、1012Or 1013cfu or total cell number. In another aspect, a therapeutic composition comprises up to about 105、106、107、108、109、1010、1011、1012、1013Or 1014cfu or total cell number.

In another aspect, a therapeutic composition comprises at least about 105、106、107、108、109、1010、1011、1012Or 1013Individual cell or total cell number. In another aspect, a therapeutic composition comprises up to about 105、106、107、108、109、1010、1011、1012、1013Or 1014Individual cell or total cell number.

In one aspect, the therapeutic composition is formulated as an oral capsule, microcapsule, tablet or pill. In another aspect, the capsule, microcapsule, tablet or pill is suitable for enteral delivery. In another aspect, the capsule, microcapsule, tablet or pill is an enteric capsule, microcapsule, tablet or pill. In another aspect, the capsule, microcapsule, tablet or pill has enteric solubility, acid resistance or both.

In one aspect, the present application provides the following embodiments:

38页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:大疱性表皮松解症的治疗剂

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!

技术分类